-Author name in bold denotes the presenting author
-Asterisk * with author name denotes a Non-ASH member
Clinically Relevant Abstract denotes an abstract that is clinically relevant.

PhD Trainee denotes that this is a recommended PHD Trainee Session.

Ticketed Session denotes that this is a ticketed session.

283 Robust Discovery of Candidate DNA Methylation Cancer Drivers

Program: Oral and Poster Abstracts
Type: Oral
Session: 621. Lymphoma—Genetic/Epigenetic Biology: Genetic and epigenetic profiling of malignant lymphomas
Hematology Disease Topics & Pathways:
Leukemia, Adult, Diseases, CLL, Biological Processes, epigenetics, Lymphoid Malignancies
Saturday, December 5, 2020: 3:15 PM

Heng Pan, PhD1,2,3*, Loïc Renaud, MD, MSc4,5,6,7*, Ronan Chaligne, PhD5,6,7*, Johannes Bloehdorn, MD8*, Eugen Tausch, MD8*, Daniel Mertens, PhD9*, Anna-Maria Fink, MD10*, Kirsten Fischer, MD10*, Chao Zhang, PhD3,7*, Doron Betel, PhD3,7*, Andreas Gnirke11*, Marcin Imielinski1,3,5,6,12*, Jerome Moreaux, PhD13,14,15,16, Michael Hallek10, Alexander Meissner, PhD11,17*, Stephan Stilgenbauer, MD8, Catherine J. Wu, MD11,18, Olivier Elemento, PhD1,2,3,6* and Dan A. Landau, MD, PhD3,5,6,7

1Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY
2Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY
3Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY
4Service des Maladies du Sang, Centre Hospitalier Régional Universitaire De Lille, Lille, France
5New York Genome Center, New York, NY
6Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY
7Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY
8Department of Internal Medicine III, Ulm University, Ulm, Germany
9German Cancer Research Center (DKFZ), Heidelberg, Germany
10German CLL Study Group, and Department I of Internal Medicine, and Center of Integrated Oncology ABCD, University of Cologne, Cologne, Germany
11Broad Institute of MIT and Harvard, Cambridge, MA
12Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
13UFR Médecine, Université Montpellier, Montpellier, France
14Laboratory for Monitoring Innovative Therapies, Department of Biological Hematology, CHU Montpellier, Montpellier, France
15Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
16Institut Universitaire de France, Paris, France
17Max Planck Institute for Molecular Genetics, Berlin, Germany
18Dana-Farber Cancer Institute, Boston, MA

The field of cancer genomics has been empowered by increasingly sophisticated inference tools to distinguish driver mutations from the vastly greater number of passenger mutations. Epigenetic alterations such as promoter DNA hypermethylation have been shown to drive cancer through inactivation of tumor suppressor genes (TSGs), but growing malignant populations also accrue pervasive stochastic epigenetic changes in DNA methylation (DNAme), most of which likely carry little functional impact. Unlike with somatic mutations, we have limited ability to robustly differentiate driver DNAme changes (DNAme drivers) from stochastic, passenger DNAme changes. To address this challenge, we developed MethSig, a statistical inference framework that accounts for the varying stochastic hypermethylation rates across the genome and between samples. MethSig estimates expected background DNAme changes, thereby allowing the identification of epigenetically disrupted loci, where observed hypermethylation significantly exceeds expectation, potentially reflecting positive selection (Fig. 1a).

We applied MethSig to reduced representation bisulfite sequencing (RRBS) data of chronic lymphocytic leukemia (CLL) cohorts, which include 304 CLLs collected in a prospective clinical trial (CLL8) and 103 CLLs in a previously published study (CLL-DFCI, Landau et al., 2014), as well as other malignancies where RRBS data is available, including ductal carcinoma in situ (Abba et al., 2015) and multiple myeloma. Area under the receiver operating characteristic curve (AUROC) was used to evaluate sensitivity and specificity of methods in the inference of likely DNAme drivers. We identified two key features that are likely to be strongly associated with true candidate DNAme drivers: gene silencing in relation to promoter hypermethylation and association with clinical outcome. MethSig qualitatively improved ROC across those clinical and biological read outs (0.955 of MethSig, 95% confidence interval [CI] 0.945 - 0.965, versus 0.703 of benchmarked methods, 95% CI 0.669 - 0.737, Fig. 1b used CLL8 as an example).

We identified 189 candidate DNAme drivers in CLL, which include known TSGs, and are enriched in genes hypermethylated or inactivated across cancer types. To further validate MethSig’s inferences, selected CLL candidate DNAme drivers (DUSP22, RPRM) underwent CRISPR/Cas9 knockout (KO) in CLL cells and stable KO clones were generated through single-cell cloning to eliminate genetic heterogeneity effect. The RPRM and DUSP22 KO clones showed faster growth without treatment (Fig. 1c) and superior fitness in ibrutinib/fludarabine treatment compared with controls (Fig. 1d). Notably, we observed a gene dose effect in the RPRM KO clones (Fig. 1c-d, greater growth of the bi-allelic compared to mono-allelic KO).

Elastic net regression with a Cox proportional hazards model was used to evaluate DNAme drivers’ contribution to the prediction of failure-free survival after treatment (FFS; failure defined as retreatment or death) and a rigorous training (CLL8) and validation (an independent cohort CLL-DFCI) cohort study design was implemented to safeguard from overfitting and poor generalizability. DNAme drivers were found to be associated with shorter FFS in independent CLL cohorts (Fig. 1e-f). A regression model including established CLL risk indicators (IGHV unmutated status, del[17p] or TP53 mutation) showed an adjusted hazard ratio of 2.3 (95% CI 1.6 - 3.3, P = 2 × 10-6) in CLL8 cohort and 3.2 (95% CI 1.2 - 8.8, P = 0.02) in CLL-DFCI cohort for patients with high risk. Application of MethSig to CLL relapsed after chemoimmunotherapy further identified relapse-specific DNAme drivers, enriched in TP53 targets as well as DNA damage pathway, which indicates that CLL relapse after chemotherapy may follow an alternative path compared to CLL progression in the absence of therapy, offering novel insights for therapeutic strategies to address drug-resistant or relapsed cancer.

Collectively, our data support a novel framework for the analysis of DNAme changes in cancer to specifically identify DNAme drivers of disease progression and relapse, empowering the discovery of epigenetic mechanisms that enhance cancer cell fitness. This work addressed a central gap between cancer epigenetics and cancer genetics, where such tools have had a transformative impact in precision oncology and cancer gene discovery.

Disclosures: Tausch: Janssen-Cilag: Consultancy, Honoraria, Research Funding; Roche: Consultancy, Honoraria, Research Funding; AbbVie: Consultancy, Honoraria, Research Funding. Fink: AbbVie: Other: travel grants; Janssen: Honoraria; Celgene: Research Funding. Fischer: F. Hoffmann-La Roche: Honoraria, Other: travel grants; AbbVie: Honoraria. Gnirke: FL67 Inc.: Consultancy. Moreaux: Diag2Tec: Consultancy. Hallek: Celgene: Consultancy, Research Funding; Pharmacyclics: Consultancy, Research Funding; AbbVie: Consultancy, Research Funding; Gilead: Consultancy, Research Funding; Mundipharma: Consultancy, Research Funding; Janssen: Consultancy, Research Funding; Roche: Consultancy, Research Funding. Stilgenbauer: Janssen-Cilag: Consultancy, Honoraria, Other: travel support, Research Funding; Genentech: Consultancy, Honoraria, Other: travel support, Research Funding; Gilead: Consultancy, Honoraria, Other: travel support, Research Funding; Genzyme: Consultancy, Honoraria, Other: travel support, Research Funding; Pharmacyclics: Consultancy, Honoraria, Other, Research Funding; F. Hoffmann-LaRoche: Consultancy, Honoraria, Other: travel support, Research Funding; Celgene: Consultancy, Honoraria, Other: travel support, Research Funding; Boehringer-Ingelheim: Consultancy, Honoraria, Other: travel support, Research Funding; Amgen: Consultancy, Honoraria, Other: travel support, Research Funding; AbbVie: Consultancy, Honoraria, Other: travel support, Research Funding; GlaxoSmithKline: Consultancy, Honoraria, Other: travel support, Research Funding; Novartis: Consultancy, Honoraria, Other, Research Funding; Mundipharma: Consultancy, Honoraria, Other, Research Funding. Wu: Pharmacyclics: Research Funding; BionTech: Current equity holder in publicly-traded company. Elemento: Acuamark: Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees; OneThree Biotech: Current equity holder in private company, Other: Cofounder; Owkin: Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees; Sanofi: Research Funding; Eli Lilly: Research Funding; Volastra Therapeutics: Current equity holder in private company, Other: Cofounder; Freenome: Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees; Genetic Intelligence: Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees; Janssen: Research Funding. Landau: Bristol Myers Squibb: Research Funding; Illumina: Research Funding.

<< Previous Abstract | Next Abstract
*signifies non-member of ASH